1
|
Dreschers S, Platen C, Oppermann L, Doughty C, Ludwig A, Babendreyer A, Orlikowsky TW. EGF-Receptor against Amphiregulin (AREG) Influences Costimulatory Molecules on Monocytes and T Cells and Modulates T-Cell Responses. J Immunol Res 2023; 2023:8883045. [PMID: 38046264 PMCID: PMC10691888 DOI: 10.1155/2023/8883045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Amphiregulin (AREG) is a ligand of the epidermal growth factor receptor (EGFR) and has been shown to regulate the phagocytosis-induced cell death of monocytes in peripheral blood. AREG-dependent apoptotic signaling engages factors of the intrinsic and extrinsic apoptotic pathway, such as BCL-2, BCL-XL, and death ligand/receptor CD95/CD95L. Here, we tested the hypothesis that AREG influences costimulatory monocyte functions, which are crucial for T-cell responses. We found a stronger expression of AREG and EGFR in monocytes compared to lymphocytes. As a novel function of AREG, we observed reduced T-cell proliferation following polyclonal T-cell stimulation with OKT3. This reduction of proliferation occurred in the presence of monocytes as well as in their absence, monocyte signaling being replaced by crosslinking of OKT3. Increasing concentrations of AREG down-modulated the concentration of costimulatory B7 molecules (CD80/CD86) and HLA-DR on monocytes. In proliferation assays, CD28 expression on T cells was down-modulated on the application of OKT3 but unaltered by AREG. LcK activation, following OKT3-stimulation, was reduced in T cells that had been coincubated with AREG. The effects of AREG on T-cell phenotypes were also present when monocytes were depleted and OKT3 was crosslinked. The rearranged expression of immunological synapse proteins was accompanied by an alteration of T-cell polarization. Although the proportion of regulatory T cells was not shifted by AREG, IL-17-expressing T cells were significantly enhanced, with a bias toward TH1-polarization. Taken together, these results suggest that AREG acts as an immunoregulatory molecule at the interface between antigen-presenting cells and T cells.
Collapse
Affiliation(s)
- Stephan Dreschers
- Department of Neonatology, University Children's Hospital, Aachen, Germany
| | - Christopher Platen
- Department of Neonatology, University Children's Hospital, Aachen, Germany
| | - Louise Oppermann
- Department of Neonatology, University Children's Hospital, Aachen, Germany
| | - Caitlin Doughty
- Department of Neonatology, University Children's Hospital, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
2
|
Corrêa-Moreira D, dos Santos A, Menezes RC, Morgado FN, Borba CM, Oliveira-Ferreira J. Skin Immune Response of Immunocompetent and Immunosuppressed C57BL/6 Mice After Experimental Subcutaneous Infection Caused by Purpureocillium lilacinum. Front Microbiol 2021; 12:615383. [PMID: 34194400 PMCID: PMC8236815 DOI: 10.3389/fmicb.2021.615383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Hyalohyphomycosis is a fungal infection characterized by the presence of a hyaline mycelium in the host. It is caused by several agents, such as Purpureocillium lilacinum. Our study aimed to evaluate some cell subsets and inflammatory markers involved in the in situ immune response to subcutaneous hyalohyphomycosis by P. lilacinum in C57BL/6 murine models. The fungal isolate was inoculated in mice randomly distributed in immunocompetent/infected (CI) and immunosuppressed/infected (SI) groups. Mice were evaluated on days 1, 3, 5, and 7 after inoculation. Histopathological studies showed several lesions in the site of infection as well as the formation of multifocal and mixed inflammatory infiltrates, which differed between the CI and SI groups. This analysis also revealed conidia and hypha-like structures in subcutaneous tissues of mice of both groups. The immunohistochemical analysis showed lower percentages of macrophages and neutrophils in the SI group compared to those in the CI group. Moreover, the intensity of interleukin (IL)-1β and nitric oxide synthase 2 production by cells of immunosuppressed mice was discreet, compared to immunocompetent mice that ranged from moderate to intense over time. The quantitative interference of dexamethasone in the response to the fungus was also demonstrated. We concluded that our results can be useful not only to broaden the knowledge on P. lilacinum but also, based on this host-parasite relationship, to contribute to the understanding of the mechanisms of infection.
Collapse
Affiliation(s)
- Danielly Corrêa-Moreira
- Laboratory of Taxonomy, Biochemistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Arethuza dos Santos
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Rodrigo C. Menezes
- Laboratory of Clinical Research in Dermatozoonosis in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fernanda N. Morgado
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cintia M. Borba
- Laboratory of Taxonomy, Biochemistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Joseli Oliveira-Ferreira
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Hwang JW, Myeong SH, Lee NH, Kim H, Son HJ, Chang JW, Lee NK, Na DL. Immunosuppressant Drugs Mitigate Immune Responses Generated by Human Mesenchymal Stem Cells Transplanted into the Mouse Parenchyma. Cell Transplant 2021; 30:9636897211019025. [PMID: 34044601 PMCID: PMC8168027 DOI: 10.1177/09636897211019025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
It has been widely accepted that mesenchymal stem cells (MSCs) can evade the immune surveillance of the recipient. However, emerging research cast doubt on whether MSCs are intrinsically immune-privileged. Previously, we observed that the transplantation of human MSCs (hMSCs) into the mouse parenchyma attracted a high infiltration of leukocytes into the injection tract. Thus, in order to reduce the immune responses generated by hMSCs, the aim of this study was to assess which immunosuppressant condition (dexamethasone only, tacrolimus only, or dexamethasone and tacrolimus together) would not only reduce the overall immune response but also enhance the persistence of MSCs engrafted into the caudate putamen of wild-type C57BL/6 mice. According to immunohistochemical analysis, compared to the hMSC only group, the administration of immunosuppressants (for all three conditions) reduced the infiltration of CD45-positive leukocytes and neutrophils at the site of injection. The highest hMSC persistence was detected from the group that received combinatorial administrations of dexamethasone and tacrolimus. Moreover, compared to the immunocompetent WT mouse, higher MSC engraftment was observed from the immunodeficient BALB/c mice. The results of this study support the use of immunosuppressants to tackle MSC-mediated immune responses and to possibly prolong the engraftment of transplanted MSCs.
Collapse
Affiliation(s)
- Jung Won Hwang
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Su Hyeon Myeong
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Na-Hee Lee
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea
| | - Hyeongseop Kim
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell Institute, ENCell Co. Ltd., Seoul, Republic of Korea
| | - Hyo Jin Son
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea.,School of Medicine, 35019Sungkyunkwan University, Seoul, Republic of Korea
| | - Jong Wook Chang
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell Institute, ENCell Co. Ltd., Seoul, Republic of Korea
| | - Na Kyung Lee
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea.,School of Medicine, 35019Sungkyunkwan University, Seoul, Republic of Korea
| | - Duk L Na
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea.,School of Medicine, 35019Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Dangi A, Yu S, Lee FT, Burnette M, Wang JJ, Kanwar YS, Zhang ZJ, Abecassis M, Thorp EB, Luo X. Murine cytomegalovirus dissemination but not reactivation in donor-positive/recipient-negative allogeneic kidney transplantation can be effectively prevented by transplant immune tolerance. Kidney Int 2020; 98:147-158. [PMID: 32471635 PMCID: PMC7311252 DOI: 10.1016/j.kint.2020.01.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/17/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
Cytomegalovirus (CMV) reactivation from latently infected donor organs post-transplantation and its dissemination cause significant comorbidities in transplant recipients. Transplant-induced inflammation combined with chronic immunosuppression has been thought to provoke CMV reactivation and dissemination, although sequential events in this process have not been studied. Here, we investigated this process in a high-risk donor CMV-positive to recipient CMV-negative allogeneic murine kidney transplantation model. Recipients were either treated with indefinite immunosuppression or tolerized in a donor-specific manner. Untreated recipients served as controls. Kidney allografts from both immunosuppressed and tolerized recipients showed minimal alloimmunity-mediated graft inflammation and normal function for up to day 60 post-transplantation. However, despite the absence of such inflammation in the immunosuppressed and tolerized groups, CMV reactivation in the donor positive kidney allograft was readily observed. Interestingly, subsequent CMV replication and dissemination to distant organs only occurred in immunosuppressed recipients in which CMV-specific CD8 T cells were functionally impaired; whereas in tolerized recipients, host anti-viral immunity was well-preserved and CMV dissemination was effectively prevented. Thus, our studies uncoupled CMV reactivation from its dissemination, and underscore the potential role of robust transplantation tolerance in preventing CMV diseases following allogeneic kidney transplantation.
Collapse
Affiliation(s)
- Anil Dangi
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Shuangjin Yu
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Division of Organ Transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Frances T Lee
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Melanie Burnette
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Jiao-Jing Wang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yashpal S Kanwar
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zheng J Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael Abecassis
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Xunrong Luo
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
5
|
Dantoft W, Martínez-Vicente P, Jafali J, Pérez-Martínez L, Martin K, Kotzamanis K, Craigon M, Auer M, Young NT, Walsh P, Marchant A, Angulo A, Forster T, Ghazal P. Genomic Programming of Human Neonatal Dendritic Cells in Congenital Systemic and In Vitro Cytomegalovirus Infection Reveal Plastic and Robust Immune Pathway Biology Responses. Front Immunol 2017; 8:1146. [PMID: 28993767 PMCID: PMC5622154 DOI: 10.3389/fimmu.2017.01146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
Neonates and especially premature infants are highly susceptible to infection but still can have a remarkable resilience that is poorly understood. The view that neonates have an incomplete or deficient immune system is changing. Human neonatal studies are challenging, and elucidating host protective responses and underlying cognate pathway biology, in the context of viral infection in early life, remains to be fully explored. In both resource rich and poor settings, human cytomegalovirus (HCMV) is the most common cause of congenital infection. By using unbiased systems analyses of transcriptomic resources for HCMV neonatal infection, we find the systemic response of a preterm congenital HCMV infection, involves a focused IFN regulatory response associated with dendritic cells. Further analysis of transcriptional-programming of neonatal dendritic cells in response to HCMV infection in culture revealed an early dominant IFN-chemokine regulatory subnetworks, and at later times the plasticity of pathways implicated in cell-cycle control and lipid metabolism. Further, we identify previously unknown suppressed networks associated with infection, including a select group of GPCRs. Functional siRNA viral growth screen targeting 516-GPCRs and subsequent validation identified novel GPCR-dependent antiviral (ADORA1) and proviral (GPR146, RGS16, PTAFR, SCTR, GPR84, GPR85, NMUR2, FZ10, RDS, CCL17, and SORT1) roles. By contrast a gene family cluster of protocadherins is significantly differentially induced in neonatal cells, suggestive of possible immunomodulatory roles. Unexpectedly, programming responses of adult and neonatal dendritic cells, upon HCMV infection, demonstrated comparable quantitative and qualitative responses showing that functionally, neonatal dendritic cell are not overly compromised. However, a delay in responses of neonatal cells for IFN subnetworks in comparison with adult-derived cells are notable, suggestive of subtle plasticity differences. These findings support a set-point control mechanism rather than immaturity for explaining not only neonatal susceptibility but also resilience to infection. In summary, our findings show that neonatal HCMV infection leads to a highly plastic and functional robust programming of dendritic cells in vivo and in vitro. In comparison with adults, a minimal number of subtle quantitative and temporal differences may contribute to variability in host susceptibility and resilience, in a context dependent manner.
Collapse
Affiliation(s)
- Widad Dantoft
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Pablo Martínez-Vicente
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - James Jafali
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lara Pérez-Martínez
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Quantitative Proteomics, Institute of Molecular Biology, Mainz, Germany
| | - Kim Martin
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Synexa Life Sciences, Cape Town, South Africa
| | - Konstantinos Kotzamanis
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie Craigon
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Manfred Auer
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,SynthSys-Centre for Synthetic and Systems Biology, School of Engineering, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil T Young
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Paul Walsh
- NSilico Life Science and Department of Computing, Institute of Technology, Cork, Ireland
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Thorsten Forster
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Ghazal
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Dreschers S, Saupp P, Hornef M, Prehn A, Platen C, Morschhäuser J, Orlikowsky TW. Reduced PICD in Monocytes Mounts Altered Neonate Immune Response to Candida albicans. PLoS One 2016; 11:e0166648. [PMID: 27870876 PMCID: PMC5117704 DOI: 10.1371/journal.pone.0166648] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/01/2016] [Indexed: 01/06/2023] Open
Abstract
Background Invasive fungal infections with Candida albicans (C. albicans) occur frequently in extremely low birthweight (ELBW) infants and are associated with poor outcome. Phagocytosis of C.albicans initializes apoptosis in monocytes (phagocytosis induced cell death, PICD). PICD is reduced in neonatal cord blood monocytes (CBMO). Hypothesis Phagocytosis of C. albicans causes PICD which differs between neonatal monocytes (CBMO) and adult peripheral blood monocytes (PBMO) due to lower stimulation of TLR-mediated immune responses. Methods The ability to phagocytose C. albicans, expression of TLRs, the induction of apoptosis (assessment of sub-G1 and nick-strand breaks) were analyzed by FACS. TLR signalling was induced by agonists such as lipopolysaccharide (LPS), Pam3Cys, FSL-1 and Zymosan and blocked (neutralizing TLR2 antibodies and MYD88 inhibitor). Results Phagocytic indices of PBMO and CBMO were similar. Following stimulation with agonists and C. albicans induced up-regulation of TLR2 and consecutive phosphorylation of MAP kinase P38 and expression of TNF-α, which were stronger on PBMO compared to CBMO (p < 0.005). Downstream, TLR2 signalling initiated caspase-3-dependent PICD which was found reduced in CBMO (p < 0.05 vs PBMO). Conclusion Our data suggest direct involvement of TLR2-signalling in C. albicans-induced PICD in monocytes and an alteration of this pathway in CBMO.
Collapse
Affiliation(s)
- Stephan Dreschers
- Department of Neonatology, University Children’s Hospital, Aachen, Germany
| | - Peter Saupp
- Department of Neonatology, University Children’s Hospital, Aachen, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology, University Children’s Hospital, Aachen, Germany
| | - Andrea Prehn
- Department of Environmental Medicine, University Children’s Hospital, Aachen, Germany
| | - Christopher Platen
- Department of Neonatology, University Children’s Hospital, Aachen, Germany
| | | | | |
Collapse
|
7
|
Im SA, Gerelchuluun T, Lee CK. Evidence for Direct Inhibition of MHC-Restricted Antigen Processing by Dexamethasone. Immune Netw 2014; 14:328-32. [PMID: 25550699 PMCID: PMC4275390 DOI: 10.4110/in.2014.14.6.328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/24/2014] [Accepted: 12/05/2014] [Indexed: 01/08/2023] Open
Abstract
Dexamethasone (Dex) was shown to inhibit the differentiation, maturation, and antigen-presenting function of dendritic cells (DC) when added during DC generation or maturation stages. Here, we examined the direct effects of Dex on MHC-restricted antigen processing. Macrophages were incubated with microencapsulated ovalbumin (OVA) in the presence of different concentrations of Dex for 2 h, and the efficacy of OVA peptide presentation was evaluated using OVA-specific CD8 and CD4 T cells. Dex inhibited both class I- and class II-restricted presentation of OVA to T cells; this inhibitory effect on antigen presentation was much more potent in immature macrophages than in mature macrophages. The presentation of the exogenously added OVA peptide SIINFEKL was not blocked by Dex. In addition, short-term treatment of macrophages with Dex had no discernible effects on the phagocytic activity, total expression levels of MHC molecules or co-stimulatory molecules. These results demonstrate that Dex inhibits intracellular processing events of phagocytosed antigens in macrophages.
Collapse
Affiliation(s)
- Sun-A Im
- College of Pharmacy, Chungbuk National University, Cheongju 362-763, Korea
| | - Turmunkh Gerelchuluun
- Deaprtment of Biotechnology and Nutrition School of Industrial Technology, Mongolian University of Science and Technology, Ulaanbaatar, 210646, Mongolia
| | - Chong-Kil Lee
- College of Pharmacy, Chungbuk National University, Cheongju 362-763, Korea
| |
Collapse
|
8
|
Peine KJ, Guerau-de-Arellano M, Lee P, Kanthamneni N, Severin M, Probst GD, Peng H, Yang Y, Vangundy Z, Papenfuss TL, Lovett-Racke AE, Bachelder EM, Ainslie KM. Treatment of experimental autoimmune encephalomyelitis by codelivery of disease associated Peptide and dexamethasone in acetalated dextran microparticles. Mol Pharm 2014; 11:828-35. [PMID: 24433027 PMCID: PMC3993881 DOI: 10.1021/mp4005172] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system that can cause loss of motor function and is thought to result, in part, from chronic inflammation due to an antigen-specific T cell immune response. Current treatments suppress the immune system without antigen specificity, increasing the risks of cancer, chronic infection, and other long-term side effects. In this study, we show treatment of experimental autoimmune encephalomyelitis (EAE), a model of MS, by coencapsulating the immunodominant peptide of myelin oligodendrocyte glycoprotein (MOG) with dexamethasone (DXM) into acetalated dextran (Ac-DEX) microparticles (DXM/MOG/MPs) and administering the microparticles subcutaneously. The clinical score of the mice was reduced from 3.4 to 1.6 after 3 injections 3 days apart with the coencapsulated microparticulate formulation (MOG 17.6 μg and DXM 8 μg). This change in clinical score was significantly greater than observed with phosphate-buffered saline (PBS), empty MPs, free DXM and MOG, DXM/MPs, and MOG/MPs. Additionally, treatment with DXM/MOG/MPs significantly inhibited disease-associated cytokine (e.g., IL-17, GM-CSF) expression in splenocytes isolated in treated mice. Here we show a promising approach for the therapeutic treatment of MS using a polymer-based microparticle delivery platform.
Collapse
Affiliation(s)
- Kevin J Peine
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University , Columbus, Ohio, 43210, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Gille C, Orlikowsky TW, Spring B, Hartwig UF, Wilhelm A, Wirth A, Goecke B, Handgretinger R, Poets CF, André MC. Monocytes derived from humanized neonatal NOD/SCID/IL2Rγ(null) mice are phenotypically immature and exhibit functional impairments. Hum Immunol 2012; 73:346-54. [PMID: 22330087 DOI: 10.1016/j.humimm.2012.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 12/23/2011] [Accepted: 01/24/2012] [Indexed: 02/08/2023]
Abstract
Trials of immune-modulating drugs in septic patients have mostly failed to demonstrate clinical efficacy. Thus, we sought to generate a surrogate model of myelomonocytic lineage differentiation that would potentially allow sepsis induction and preclinical testing of anti-inflammatory drugs. Comparing transplantation of cord blood-derived stem cells in neonatal NOD/SCID/IL2Rγ(null) (neonatal huNSG) mice with transplantation of adult peripheral mobilized stem cells into adult NSG (adult huNSG) recipients, we demonstrate that myelomonocytic lineage differentiation in neonatal huNSG mice is retarded and monocytes are phenotypically immature with respect to HLA-DR expression and the emergence of CD80(+)CD86(+) monocytes. Functionally, neonatal huNSG mice were less sensitive toward interferon-γ-induced upregulation of CD86 and exhibited a reduced T-cell stimulating capacity when compared with adult huNSG mice, whereas the phagocytic activity and the ability for cytokine secretion were mature. However, comparison of these data with data obtained from human neonates indicate that absence of the CD80(+)CD86(+) population and the reduced T-cell stimulating capacity of neonatal huNSG monocytes resemble functional immaturities observed in human neonatal monocytes. Thus, these two mouse models might well serve as 2 independent surrogate models for studying the neonatal myelomonocytic lineage differentiation or for testing the efficacy of immunomodulatory drugs on functionally mature monocytes.
Collapse
Affiliation(s)
- Christian Gille
- University Children's Hospital, Department of Neonatology, Eberhard Karls University, 72076 Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Gille C, Dreschers S, Spring B, Tárnok A, Bocsi J, Poets CF, Orlikowsky TW. Differential modulation of cord blood and peripheral blood monocytes by intravenous immunoglobulin. CYTOMETRY PART B-CLINICAL CYTOMETRY 2011; 82:26-34. [PMID: 21812105 DOI: 10.1002/cyto.b.20609] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 04/06/2011] [Accepted: 05/26/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND Immunoglobulins (IVIG) have been shown to be useful in adults suffering from sepsis. In contrast, prophylactic and curative IVIG trials failed to show beneficial effects in neonates. We tested the hypothesis that IVIG, have different effects on monocytes from cord blood (CBMO) and peripheral blood monocytes from adults (PBMO) with respect to survival, phenotype, and function. METHODS Mononuclear cells, or purified monocytes, were cultured in 5% human serum, incubated with polyvalent IVIG (1 mg/ml), stimulated with green fluorescent protein (GFP)-labeled Escherichia coli (E. Coli-GFP), Interferon-γ (IFN-γ, 50 U/ml), or the T cell mitogen anti-CD3 monoclonal antibody, αCD3-mAb, (5 μg/ml). Phagocytosis, phenotype, T cell proliferation, and apoptosis were assessed by flow cytometry. RESULTS IVIG enhanced phagocytosis in PBMO or CBMO when infected directly after isolation, while IVIG had no effect on monocytes cultured 48 h prior to infection. In contrast to PBMO, IVIG inhibited the IFN-γ mediated up-regulation of CD80, CD86, and HLA-DR on CBMO. In the presence of IVIG, stimulation with αCD3 in cord blood enhanced deletion, inhibited blast formation and CD28 up-regulation of T cells (P < 0.05 vs. T cells from adults). IVIG induced monocyte apoptosis, associated with up-regulation of Annexin V and loss of nuclear DNA, which was more pronounced in CBMO. Although phagocytosis induced cell death (PICD) was lower in CBMO (P < 0.05 vs. PBMO), the addition of IVIG enhanced PICD levels of CBMO to the extent of PBMO. CONCLUSIONS IVIG inhibits co-stimulatory receptors and functions of CBMO and induces apoptosis. These findings may be of clinical relevance for the failure of IVIG benefit in neonatal sepsis.
Collapse
Affiliation(s)
- Christian Gille
- Department of Neonatology, University Children's Hospital, Calwerstr.7, 72076 Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
11
|
Neunhoeffer F, Lipponer D, Eichner M, Poets CF, Wacker A, Orlikowsky TW. Influence of Gestational Age, Cesarean Section and Hematocrit on lnterleukin-8 Concentrations in Plasma and Detergent-Lysed Whole Blood of Noninfected Newborns. Transfus Med Hemother 2011; 38:183-189. [PMID: 21760762 PMCID: PMC3128150 DOI: 10.1159/000328860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Accepted: 11/30/2011] [Indexed: 11/19/2022] Open
Abstract
SUMMARY: BACKGROUND: Sensitivity of interleukin-8 (IL-8) in detecting early-onset bacterial infection (EOBI) is high. A high percentage is bound to nonspecific receptors on erythrocytes which can be determined via cell lysis. We have shown detergent-lysed whole blood (DLWB) IL-8 to be superior to plasma IL-8 in detecting EOBI. METHODS: To evaluate influence of pre- and perinatal factors on plasma and DLWB IL-8 concentrations, IL-8 was determined via ELISA (Immu-lite) in 146 noninfected newborns with risk factors for EOBI at two different time periods: 0-6 (group I) and 24-30 h (group II) after birth. The influence of gender, mode of delivery, gestational age and hematocrit was evaluated. RESULTS: While we found no influence of gender or gestational age, hematocrit was positively correlated with IL-8 plasma concentration (group I: r = 0.33, p < 0.001; group II: r = 0.30, p <0.01). IL-8 plasma concentrations after primary versus secondary cesarean section were lower (p < 0.05). Gestational age was correlated with DLWB IL-8 concentrations (group I: r = 0.46, p < 0.001; group II: r = 0.28, p < 0.001). CONCLUSION: Plasma IL-8 concentrations were positively correlated with hematocrit, whereas DLWB IL-8 concentrations increased with gestational age. This may be relevant to the interpretation of IL-8 in preterm infants and infants with anemia, polyglobulia or hematolytic diseases.
Collapse
Affiliation(s)
| | - Diana Lipponer
- Department of Neonatology, University Children's Hospital, Germany
| | - Martin Eichner
- Department of Medical Biometry, University of Tubingen, Germany
| | | | - Annette Wacker
- Department of Neonatology, University Children's Hospital, Germany
| | | |
Collapse
|
12
|
Gnade BT, Moen ST, Chopra AK, Peterson JW, Yeager LA. Emergence of anthrax edema toxin as a master manipulator of macrophage and B cell functions. Toxins (Basel) 2010; 2:1881-97. [PMID: 22069663 PMCID: PMC3153274 DOI: 10.3390/toxins2071881] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 07/06/2010] [Accepted: 07/12/2010] [Indexed: 11/19/2022] Open
Abstract
Anthrax edema toxin (ET), a powerful adenylyl cyclase, is an important virulence factor of Bacillus anthracis. Until recently, only a modest amount of research was performed to understand the role this toxin plays in the organism's immune evasion strategy. A new wave of studies have begun to elucidate the effects this toxin has on a variety of host cells. While efforts have been made to illuminate the effect ET has on cells of the adaptive immune system, such as T cells, the greatest focus has been on cells of the innate immune system, particularly the macrophage. Here we discuss the immunoevasive activities that ET exerts on macrophages, as well as new research on the effects of this toxin on B cells.
Collapse
Affiliation(s)
- Bryan T. Gnade
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (B.T.G.); (S.T.M.)
| | - Scott T. Moen
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (B.T.G.); (S.T.M.)
| | - Ashok K. Chopra
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (B.T.G.); (S.T.M.)
- Center for Biodefense and Emerging Infectious Diseases and Sealy Center for Vaccine Development, The University of Texas Medical Branch, Galveston, TX 77555, USA; (A.K.C.); (J.W.P.)
| | - Johnny W. Peterson
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (B.T.G.); (S.T.M.)
- Center for Biodefense and Emerging Infectious Diseases and Sealy Center for Vaccine Development, The University of Texas Medical Branch, Galveston, TX 77555, USA; (A.K.C.); (J.W.P.)
| | - Linsey A. Yeager
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (B.T.G.); (S.T.M.)
| |
Collapse
|
13
|
Dudhgaonkar SP, Janardhanam SB, Kodumudi KN, Srinivasan M. CD80 blockade enhance glucocorticoid-induced leucine zipper expression and suppress experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2009; 183:7505-13. [PMID: 19917693 DOI: 10.4049/jimmunol.0902056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Designing mimetic of the interface functional groups of known receptor-ligand complexes is an attractive strategy for developing potential therapeutic agents that interfere with target protein-protein interactions. The CD80/CD86-CD28/CD152 costimulatory interactions transmit signals for CD4(+) T cell activation and suppression and are critically involved in the initiation, progression, and reactivation of the immunopathology in multiple sclerosis. Differences in the pattern, levels, and kinetics of expression of CD80/CD86 molecules in conjunction with differences in the strength of the signals delivered upon binding CD28 or CD152 determine the outcome of the immune response. A temporal up-regulation of surface expression of CD80 relative to CD86 on APCs and CNS-infiltrating cells has been shown to correlate with disease progression in experimental autoimmune encephalomyelitis an animal model for multiple sclerosis. Hence blockade of the CD80 costimulatory axis has therapeutic potential in multiple sclerosis. In this study, we report the efficacy of a novel CD80-blocking agent CD80-competitive antagonist peptide (CD80-CAP) in suppressing clinical disease and relapse in experimental autoimmune encephalomyelitis. The CD80-CAP mediates protection by inhibiting proinflammatory cytokines and skewing toward anti-inflammatory response presumably by enhancing the expression of glucocorticoid-induced leucine zipper in activated CD4(+) T cells.
Collapse
Affiliation(s)
- Shailesh P Dudhgaonkar
- Department of Oral Pathology, Medicine and Radiology, School of Dentistry, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
14
|
Gille C, Leiber A, Spring B, Kempf VAJ, Loeffler J, Poets CF, Orlikowsky TW. Diminished phagocytosis-induced cell death (PICD) in neonatal monocytes upon infection with Escherichia coli. Pediatr Res 2008; 63:33-8. [PMID: 18043500 DOI: 10.1203/pdr.0b013e31815b8e9f] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
An imbalance in apoptosis or survival of immune cells plays an essential role in the pathophysiology of sepsis. Phagocytosis-induced cell death (PICD) is a common result of the pathogen-host cell interaction mediated by reactive oxygen species (ROS). Neonatal sepsis is frequently characterized by hyperinflammation. Cord blood monocytes (CBMO) are equivalent to monocytes of adults [peripheral blood monocytes (PBMO)], both in terms of phagocytosis and killing of Escherichia coli. We investigated whether CBMO are less sensitive toward PICD compared with PBMO. Monocytes were infected with green fluorescent protein (GFP)-labeled E. coli. Phagocytic activity, cell-count, Annexin V staining, hypoploid DNA content, CD95 and CD95L expression, and caspase-8 and -9 activities were analyzed by flow cytometry, ROS production by chemiluminescence, and CD95L mRNA expression by reverse-transcriptase polymerase chain reaction. With equal phagocytic activity and ROS production, PBMO cell count was decreased by 82 +/- 6% versus 28 +/- 8% for CBMO after infection. Annexin V binding was enhanced fivefold on PBMO; 56 +/- 15% of PBMO showed a hypodiploid DNA content compared with 9 +/- 6% of CBMO. Caspases CD95L and CD95L mRNA were up-regulated in PBMO. Our results indicate that CBMO are less sensitive toward E. coli-mediated PICD than PBMO. Modifying monocyte apoptosis may be a target for future interventions in sepsis.
Collapse
Affiliation(s)
- Christian Gille
- Department of Neonatology, University Children's Hospital, Tuebingen, 72076, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Dolff S, Wilde B, Patschan S, Dürig J, Specker C, Philipp T, Kribben A, Witzke O. Peripheral Circulating Activated B-cell Populations are Associated with Nephritis and Disease Activity in Patients with Systemic Lupus Erythematosus. Scand J Immunol 2007; 66:584-90. [PMID: 17868260 DOI: 10.1111/j.1365-3083.2007.02008.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
B-cell-dependent autoantibody production is a hallmark of systemic lupus erythematosus (SLE) which requires costimulatory molecules. The aim of the study was to analyse the expression of costimulatory molecules on B cells in patients with SLE. Twenty-six patients with SLE (four male, 22 female, mean age 46 +/- 15 years) as defined by the American College of Rheumatology criteria and 13 healthy controls (three male, 10 female, mean age 43 +/- 15 years) were included in the study. In a subgroup analysis, SLE patients were divided according to renal involvement due to SLE (10 with and 16 patients without renal involvement). Clinical disease activity was assessed according to the systemic lupus erythematosus disease activity index (SLEDAI). Blood B-cell populations were analysed by FACS for the cell surface marker expression of CD27, CD38, CD71, CD80, CD86 and CD137 ligand. The expression levels of CD71, CD80 and CD86 on B cells were significantly enhanced in SLE patients when compared with healthy controls (27 +/- 3% versus 11 +/- 2%, P = 0.0003, 55 +/- 2% versus 28 +/- 4%, P < 0.0001, 34 +/- 3% versus 12 +/- 2%, P < 0.0001). CD86 expression was significantly elevated in patients with renal involvement when compared with patients without renal disease (43 +/- 6% versus 28 +/- 3%, P < 0.05). There was a significant correlation between the expression levels of CD80 and CD86 on CD19(+) B cells and disease activity. Moreover, prednisone dose significantly correlated with SLEDAI (r = 0.5, P = 0.02) and with the expression levels of CD86 (r = 0.47, P = 0.02). A pathological B-cell population is associated with disease activity and renal involvement in SLE which are obviously resistant to therapy with medium doses of prednisone.
Collapse
Affiliation(s)
- S Dolff
- Department of Nephrology, University Hospital Essen, Essen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Gille C, Orlikowsky TW. Flow Cytometric Methods in the Detection of Neonatal Infection. Transfus Med Hemother 2007. [DOI: 10.1159/000101519] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
17
|
Gille C, Spring B, Tewes LJ, Löffler J, Dannecker GE, Hoffmann MK, Eichner M, Poets CF, Orlikowsky TW. Diminished response to interleukin-10 and reduced antibody-dependent cellular cytotoxicity of cord blood monocyte-derived macrophages. Pediatr Res 2006; 60:152-7. [PMID: 16864695 DOI: 10.1203/01.pdr.0000228345.58509.7b] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Monocyte-derived macrophage (MPhi) subsets are generated by antagonistic induction pathways. A helper MPhi-type (Mh-MPhi) is induced by interferon gamma (IFN-gamma), whereas a cytotoxic MPhi-type (Mc-MPhi), induced by interleukin-10 (IL-10), is a potent mediator of antibody-dependent cellular cytotoxicity (ADCC). Compared with MPhi from healthy adults [peripheral blood monocyte-derived macrophages (PBMPhi)], cord blood MPhi (CBMPhi) were found less capable of generating Mh-MPhi. Here we tested the hypothesis that their generation of Mc-MPhi via IL-10 is also impaired. MPhi surface markers were phenotyped. IL-10 protein and mRNA production were detected after stimulation [alphaCD3 monoclonal antibody (mAb)]. CBMPhi or PBMPhi were co-cultured with MPhi-depleted mononuclear cells of adults and CD4-targeting antibodies as models for ADCC were added. In cord blood, we found diminished alphaCD3-induced IL-10 protein and mRNA production (p < 0.05 versus adults). Basal CD16 and HLA-DR expressions on CBMPhi of preterm and full-term neonates were lower (p < 0.05 versus PBMPhi). IL-10 had reduced effects on CD16 up- and HLA-DR down-modulation on CBMPhi (p < 0.05 versus PBMPhi). CD4-directed receptor modulation and deletion were reduced in the presence of CBMPhi (p < 0.05 versus PBMPhi). IL-10 failed to enhance their ADCC capacity, which was in contrast to PBMPhi (p < 0.05). These data suggest that CBMPhi have an impaired cytotoxic capacity via lower sensitivity toward IL-10.
Collapse
|